1
|
Filip CI, Cătană A, Kutasi E, Roman SA, Militaru MS, Risteiu GA, Dindelengan GC. Breast Cancer Screening and Prophylactic Mastectomy for High-Risk Women in Romania. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:570. [PMID: 38674216 PMCID: PMC11052261 DOI: 10.3390/medicina60040570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/10/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024]
Abstract
Breast cancer remains a significant contributor to morbidity and mortality within oncology. Risk factors, encompassing genetic and environmental influences, significantly contribute to its prevalence. While germline mutations, notably within the BRCA genes, are commonly associated with heightened breast cancer risk, a spectrum of other variants exists among affected individuals. Diagnosis relies on imaging techniques, biopsies, biomarkers, and genetic testing, facilitating personalised risk assessment through specific scoring systems. Breast cancer screening programs employing mammography and other imaging modalities play a crucial role in early detection and management, leading to improved outcomes for affected individuals. Regular screening enables the identification of suspicious lesions or abnormalities at earlier stages, facilitating timely intervention and potentially reducing mortality rates associated with breast cancer. Genetic mutations guide screening protocols, prophylactic interventions, treatment modalities, and patient prognosis. Prophylactic measures encompass a range of interventions, including chemoprevention, hormonal inhibition, oophorectomy, and mastectomy. Despite their efficacy in mitigating breast cancer incidence, these interventions carry potential side effects and psychological implications, necessitating comprehensive counselling tailored to individual cases.
Collapse
Affiliation(s)
- Claudiu Ioan Filip
- Department of Plastic Surgery and Burn Unit, Emergency District Hospital, 400535 Cluj-Napoca, Romania; (C.I.F.); (G.C.D.)
- First Surgical Clinic, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania
| | - Andreea Cătană
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (E.K.); (S.A.R.); (G.A.R.)
- Department of Oncogeneticcs, Institute of Oncology, “Prof. Dr. I. Chiricuță”, 400015 Cluj-Napoca, Romania
- Regional Laboratory Cluj-Napoca, Department of Medical Genetics, Regina Maria Health Network, 400363 Cluj-Napoca, Romania
| | - Eniko Kutasi
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (E.K.); (S.A.R.); (G.A.R.)
| | - Sara Alexia Roman
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (E.K.); (S.A.R.); (G.A.R.)
| | - Mariela Sanda Militaru
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (E.K.); (S.A.R.); (G.A.R.)
- Regional Laboratory Cluj-Napoca, Department of Medical Genetics, Regina Maria Health Network, 400363 Cluj-Napoca, Romania
| | - Giulia Andreea Risteiu
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (E.K.); (S.A.R.); (G.A.R.)
| | - George Călin Dindelengan
- Department of Plastic Surgery and Burn Unit, Emergency District Hospital, 400535 Cluj-Napoca, Romania; (C.I.F.); (G.C.D.)
- First Surgical Clinic, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania
| |
Collapse
|
2
|
Sarfraz Z, Sarfraz A, Mehak O, Akhund R, Bano S, Aftab H. Racial and socioeconomic disparities in triple-negative breast cancer treatment. Expert Rev Anticancer Ther 2024; 24:107-116. [PMID: 38436305 DOI: 10.1080/14737140.2024.2326575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) continues to be a significant concern, especially among minority populations, where treatment disparities are notably pronounced. Addressing these disparities, especially among African American women and other minorities, is crucial for ensuring equitable healthcare. AREAS COVERED This review delves into the continuum of TNBC treatment, noting that the standard of care, previously restricted to chemotherapy, has now expanded due to emerging clinical trial results. With advances like PARP inhibitors, immunotherapy, and antibody-drug conjugates, a more personalized treatment approach is on the horizon. The review highlights innovative interventions tailored for minorities, such as utilizing technology like text messaging, smartphone apps, and targeted radio programming, coupled with church-based behavioral interventions. EXPERT OPINION Addressing TNBC treatment disparities demands a multifaceted approach, blending advanced medical treatments with culturally sensitive community outreach. The potential of technology, especially in the realm of promoting health awareness, is yet to be fully harnessed. As the field progresses, understanding and integrating the socio-economic, biological, and access-related challenges faced by minorities will be pivotal for achieving health equity in TNBC care.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Azza Sarfraz
- Department of Pediatrics, Aga Khan University, Karachi, Pakistan
| | - Onaiza Mehak
- Department of Medicine, Aziz Fatimah Medical and Dental College, Faisalabad, Pakistan
| | - Ramsha Akhund
- Department of Surgery, University of Alabama at Birmingham, Tuscaloosa, AL, USA
| | - Shehar Bano
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Hinna Aftab
- Department of Medicine, CMH Lahore Medical College, Lahore, Pakistan
| |
Collapse
|
3
|
Skinner MK. Epigenetic biomarkers for disease susceptibility and preventative medicine. Cell Metab 2024; 36:263-277. [PMID: 38176413 DOI: 10.1016/j.cmet.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
The development of molecular biomarkers for disease makes it possible for preventative medicine approaches to be considered. Therefore, therapeutics, treatments, or clinical management can be used to delay or prevent disease development. The problem with genetic mutations as biomarkers is the low frequency with genome-wide association studies (GWASs), generally at best a 1% association of the patients with the disease. In contrast, epigenetic alterations have a high-frequency association of greater than 90%-95% of individuals with pathology in epigenome-wide association studies (EWASs). A wide variety of human diseases have been shown to have epigenetic biomarkers that are disease specific and that detect pathology susceptibility. This review is focused on the epigenetic biomarkers for disease susceptibility, and it distinct from the large literature on epigenetics of disease etiology or progression. The development of efficient epigenetic biomarkers for disease susceptibility will facilitate a paradigm shift from reactionary medicine to preventative medicine.
Collapse
Affiliation(s)
- Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA.
| |
Collapse
|
4
|
Stan DL, Kim JO, Schaid DJ, Carlson EE, Kim CA, Sinnwell JP, Couch FJ, Vachon CM, Cooke AL, Goldenberg BA, Pruthi S. Breast Cancer Polygenic-Risk Score Influence on Risk-Reducing Endocrine Therapy Use: Genetic Risk Estimate (GENRE) Trial 1-Year and 2-Year Follow-Up. Cancer Prev Res (Phila) 2024; 17:77-84. [PMID: 38154464 DOI: 10.1158/1940-6207.capr-23-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/26/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
Refinement of breast cancer risk estimates with a polygenic-risk score (PRS) may improve uptake of risk-reducing endocrine therapy (ET). A previous clinical trial assessed the influence of adding a PRS to traditional risk estimates on ET use. We stratified participants according to PRS-refined breast cancer risk and evaluated ET use and ET-related quality of life (QOL) at 1-year (previously reported) and 2-year follow-ups. Of 151 participants, 58 (38.4%) initiated ET, and 22 (14.6%) discontinued ET by 2 years; 42 (27.8%) and 36 (23.8%) participants were using ET at 1- and 2-year follow-ups, respectively. At the 2-year follow-up, 39% of participants with a lifetime breast cancer risk of 40.1% to 100.0%, 18% with a 20.1% to 40.0% risk, and 16% with a 0.0% to 20.0% risk were taking ET (overall P = 0.01). Moreover, 40% of participants whose breast cancer risk increased by 10% or greater with addition of the PRS to a traditional breast cancer-risk model were taking ET versus 0% whose risk decreased by 10% or greater (P = 0.004). QOL was similar for participants taking or not taking ET at 1- and 2-year follow-ups, although most who discontinued ET did so because of adverse effects. However, these QOL results may have been skewed by the long interval between QOL surveys and lack of baseline QOL data. PRS-informed breast cancer prevention counseling has a lasting, but waning, effect over time. Additional follow-up studies are needed to address the effect of PRS on ET adherence, ET-related QOL, supplemental breast cancer screening, and other risk-reducing behaviors. PREVENTION RELEVANCE Risk-reducing medications for breast cancer are considerably underused. Informing women at risk with precise and individualized risk assessment tools may substantially affect the incidence of breast cancer. In our study, a risk assessment tool (IBIS-polygenic-risk score) yielded promising results, with 39% of women at highest risk starting preventive medication.
Collapse
Affiliation(s)
- Daniela L Stan
- Breast Diagnostic Clinic, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota
| | - Julian O Kim
- Department of Radiation Oncology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Daniel J Schaid
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Erin E Carlson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Christina A Kim
- Department of Medical Oncology and Hematology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jason P Sinnwell
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Fergus J Couch
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Celine M Vachon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Andrew L Cooke
- Department of Radiation Oncology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Benjamin A Goldenberg
- Department of Medical Oncology and Hematology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sandhya Pruthi
- Breast Diagnostic Clinic, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
5
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
6
|
Spaeth EL, Dite GS, Hopper JL, Allman R. Validation of an Abridged Breast Cancer Risk Prediction Model for the General Population. Cancer Prev Res (Phila) 2023; 16:281-291. [PMID: 36862830 PMCID: PMC10150247 DOI: 10.1158/1940-6207.capr-22-0460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 03/04/2023]
Abstract
PREVENTION RELEVANCE In this prospective population-based cohort study, we show the improved performance of a new risk assessment model compared with a gold-standard model (BCRAT). The classification of at-risk women using this new model highlights the opportunity to improve risk stratification and implement existing clinical risk-reduction interventions.
Collapse
Affiliation(s)
| | | | - John L. Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Richard Allman
- Genetic Technologies Limited, Fitzroy, Victoria, Australia
| |
Collapse
|
7
|
Kim D, Choi I, Ha SK, Gonzalez FJ. Keratin 79 is a PPARA target that is highly expressed by liver damage. Biochem Biophys Res Commun 2023; 650:132-136. [PMID: 36796223 PMCID: PMC10681120 DOI: 10.1016/j.bbrc.2023.01.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023]
Abstract
Keratins are key structural proteins found in skin and other epithelial tissues. Keratins also protect epithelial cells from damage or stress. Fifty-four human keratins were identified and classified into two families, type I and type II. Accumulating studies showed that keratin expression is highly tissue-specific and used as a diagnostic marker for human diseases. Notably, keratin 79 (KRT79) is type II cytokeratin that was identified as regulator of hair canal morphogenesis and regeneration in skin, but its role in liver remains unclear. KRT79 is undetectable in normal mouse but its expression is significantly increased by the PPARA agonist WY-14643 and fenofibrate, and completely abolished in Ppara-null mice. The Krt79 gene has functional PPARA binding element between exon 1 and exon 2. Hepatic Krt79 is regulated by HNF4A and HER2. Moreover, hepatic KRT79 is also significantly elevated by fasting- and high-fat diet-induced stress, and these increases are completely abolished in Ppara-null mice. These findings suggest that hepatic KRT79 is controlled by PPARA and is highly associated with liver damage. Thus, KRT79 may be considered as a diagnostic marker for human liver diseases.
Collapse
Affiliation(s)
- Donghwan Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea.
| | - Inwook Choi
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Sang Keun Ha
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Allman R, Mu Y, Dite GS, Spaeth E, Hopper JL, Rosner BA. Validation of a breast cancer risk prediction model based on the key risk factors: family history, mammographic density and polygenic risk. Breast Cancer Res Treat 2023; 198:335-347. [PMID: 36749458 PMCID: PMC10020257 DOI: 10.1007/s10549-022-06834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/02/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE We compared a simple breast cancer risk prediction model, BRISK (which includes mammographic density, polygenic risk and clinical factors), against a similar model with more risk factors (simplified Rosner) and against two commonly used clinical models (Gail and IBIS). METHODS Using nested case-control data from the Nurses' Health Study, we compared the models' association, discrimination and calibration. Classification performance was compared between Gail and BRISK for 5-year risks and between IBIS and BRISK for remaining lifetime risk. RESULTS The odds ratio per standard deviation was 1.43 (95% CI 1.32, 1.55) for BRISK 5-year risk, 1.07 (95% CI 0.99, 1.14) for Gail 5-year risk, 1.72 (95% CI 1.59, 1.87) for simplified Rosner 10-year risk, 1.51 (95% CI 1.41, 1.62) for BRISK remaining lifetime risk and 1.26 (95% CI 1.16, 1.36) for IBIS remaining lifetime risk. The area under the receiver operating characteristic curve (AUC) was improved for BRISK over Gail for 5-year risk (AUC = 0.636 versus 0.511, P < 0.0001) and for BRISK over IBIS for remaining lifetime risk (AUC = 0.647 versus 0.571, P < 0.0001). BRISK was well calibrated for the estimation of both 5-year risk (expected/observed [E/O] = 1.03; 95% CI 0.73, 1.46) and remaining lifetime risk (E/O = 1.01; 95% CI 0.86, 1.17). The Gail 5-year risk (E/O = 0.85; 95% CI 0.58, 1.24) and IBIS remaining lifetime risk (E/O = 0.73; 95% CI 0.60, 0.87) were not well calibrated, with both under-estimating risk. BRISK improves classification of risk compared to Gail 5-year risk (NRI = 0.31; standard error [SE] = 0.031) and IBIS remaining lifetime risk (NRI = 0.287; SE = 0.035). CONCLUSION BRISK performs better than two commonly used clinical risk models and no worse compared to a similar model with more risk factors.
Collapse
Affiliation(s)
- Richard Allman
- Genetic Technologies Limited, 60-66 Hanover St, Fitzroy, VIC, 3065, Australia.
| | - Yi Mu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gillian S Dite
- Genetic Technologies Limited, 60-66 Hanover St, Fitzroy, VIC, 3065, Australia
| | | | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| | - Bernard A Rosner
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Galvano E, Pandit H, Sepulveda J, Ng CAS, Becher MK, Mandelblatt JS, Van Dyk K, Rebeck GW. Behavioral and transcriptomic effects of the cancer treatment tamoxifen in mice. Front Neurosci 2023; 17:1068334. [PMID: 36845433 PMCID: PMC9951777 DOI: 10.3389/fnins.2023.1068334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Tamoxifen is a common treatment for estrogen receptor-positive breast cancer. While tamoxifen treatment is generally accepted as safe, there are concerns about adverse effects on cognition. Methods We used a mouse model of chronic tamoxifen exposure to examine the effects of tamoxifen on the brain. Female C57/BL6 mice were exposed to tamoxifen or vehicle control for six weeks; brains of 15 mice were analyzed for tamoxifen levels and transcriptomic changes, and an additional 32 mice were analyzed through a battery of behavioral tests. Results Tamoxifen and its metabolite 4-OH-tamoxifen were found at higher levels in the brain than in the plasma, demonstrating the facile entry of tamoxifen into the CNS. Behaviorally, tamoxifen-exposed mice showed no impairment in assays related to general health, exploration, motor function, sensorimotor gating, and spatial learning. Tamoxifen-treated mice showed a significantly increased freezing response in a fear conditioning paradigm, but no effects on anxiety measures in the absence of stressors. RNA sequencing analysis of whole hippocampi showed tamoxifen-induced reductions in gene pathways related to microtubule function, synapse regulation, and neurogenesis. Discussion These findings of the effects of tamoxifen exposure on fear conditioning and on gene expression related to neuronal connectivity suggest that there may be CNS side effects of this common breast cancer treatment.
Collapse
Affiliation(s)
- Elena Galvano
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Harshul Pandit
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jordy Sepulveda
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Christi Anne S. Ng
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Melanie K. Becher
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jeanne S. Mandelblatt
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Kathleen Van Dyk
- Department of Psychiatry, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, United States
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
10
|
de Sousa Coelho MDPS, Pereira IC, de Oliveira KGF, Oliveira IKF, Dos Santos Rizzo M, de Oliveira VA, Carneiro da Silva FC, Torres-Leal FL, de Castro E Sousa JM. Chemopreventive and anti-tumor potential of vitamin E in preclinical breast cancer studies: A systematic review. Clin Nutr ESPEN 2023; 53:60-73. [PMID: 36657931 DOI: 10.1016/j.clnesp.2022.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Vitamin E has been investigated for its antitumor potential, including the ability to change cancer gene pathways as well as promote antioxidant and pro-oxidant activity. OBJECTIVE Therefore, this systematic review aimed to evaluate antitumor and chemopreventive activity of different vitamin E isoforms (tocopherols and tocotrienols) through in vitro and in vivo studies. METHOD The systematic review was registered in PROSPERO (No. CRD4202126207) and the search was carried out in four electronic databases (PubMed, Science Direct, Scopus and Web of Science) in June 2021 by three independent reviewers. The search equation used was: "Supplementation" AND ("Vitamin E" OR Tocopherol OR Tocotrienol) AND "breast cancer" AND (chemotherapy OR therapy OR prevention). In vitro studies and animal models of breast cancer supplemented with tocopherol or tocotrienol vitamers, alone or in combination, were included. RESULTS The results revealed 8546 relevant studies that were initially identified in our search. After analysis, a total of 12 studies were eligible for this systematic review. All studies included animal models, and 5 of them also performed in vitro experiments on cancer cell lines. The studies performed supplementation with tocopherols, mixtures (tocopherols and tocotrienols) and synthetic vitamin E forms. There was an significant association of estradiol, dendritic cells and pterostilbene in combined therapy with vitamin E. Vitamin E delayed tumor development, reduced tumor size, proliferation, viability, expression of anti-apoptotic and cell proliferation genes, and upregulated pro-apoptotic genes, tumor suppressor genes and increased immune response. The effects on oxidative stress markers and antioxidant activity were conflicting among studies. Only one study with synthetic vitamin E reported cardiotoxicity, but it did not show vitamin E genotoxicity. CONCLUSION In conclusion, vitamin E isoforms, isolated or associated, showed antitumor and chemopreventive activity. However, due to studies heterogeneity, there is a need for further analysis to establish dose, form, supplementation time and breast cancer stage.
Collapse
Affiliation(s)
- Maria do Perpetuo Socorro de Sousa Coelho
- Laboratory of Genetical Toxicology (LAPGENIC), Center for Health Sciences, Graduate Program in Pharmaceutical Sciences - Pharmaceutical Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Irislene Costa Pereira
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina, Piauí, Brazil
| | - Kynnara Gabriella Feitosa de Oliveira
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina, Piauí, Brazil
| | - Iara Katryne Fonseca Oliveira
- Department of Nutrition, Postgraduate Program in Food and Nutrition - PPGAN, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Márcia Dos Santos Rizzo
- Department of Morphology, Health Sciences Center, Federal University of Piaui, Teresina, Piauí, Brazil
| | - Victor Alves de Oliveira
- Department of Nutrition, Postgraduate Program in Food and Nutrition - PPGAN, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Francisco Leonardo Torres-Leal
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piaui, Teresina, Piauí, Brazil
| | - João Marcelo de Castro E Sousa
- Laboratory of Genetical Toxicology (LAPGENIC), Center for Health Sciences, Graduate Program in Pharmaceutical Sciences - Pharmaceutical Sciences, Federal University of Piauí, Teresina, Piauí, Brazil.
| |
Collapse
|
11
|
Dietzel F, Kolberg L, Vesper AS, Hoffmann J, Nestle-Krämling C, Zwiefel K, Friebe V, Sawicki LM, Bruckmann NM, Jannusch K, Morawitz J, Antoch G, Fehm TN, Kirchner J, Mohrmann S. Factors Influencing Residual Glandular Breast Tissue after Risk-Reducing Mastectomy in Genetically Predisposed Individuals Detected by MRI Mammography. Cancers (Basel) 2023; 15:829. [PMID: 36765786 PMCID: PMC9913581 DOI: 10.3390/cancers15030829] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
PURPOSE This study seeks to evaluate MR imaging morphological factors and other covariates that influence the presence of residual glandular tissue after risk-reducing mastectomy in patients with a familial predisposition. METHODS We analyzed women of a high-risk collective with pathogenic mutation (BRCA1 (n = 49), BRCA2 (n = 24), or further mutation (n = 9)). A total of 117 breasts were analyzed, 63 left and 54 right, from a cohort of 81 patients, who were on average 40 years old. The mean follow-up was 63 months (range 12-180 months, SD = 39.67). Retrospective analysis of MR imaging data from 2006-2022 of patients of a high-risk collective (all carriers of a pathogenic mutation) with contralateral (RRCM) or bilateral risk-reducing mastectomy (RRBM) was performed. In the image data the remaining skin flap thickness by distance measurements at eight equally distributed, clockwise points and the retromamillary area, as well as by volumetry of each breast, was elected. Residual glandular tissue was also volumetrized. In addition, patient-related covariates were recorded and their influence on postoperative residual glandular tissue and skin flap thickness was analyzed by uni- and multivariate regressions. RESULTS A significant association with postoperative residual glandular tissue was shown in multivariate analysis for the independent variables breast density, skin flap mean, and surgical method (all p-values < 0.01). A negatively significant association could be seen for the variables preoperative breast volume (p-values < 0.01) and surgeon experience (most p-values < 0.05-<0.1). CONCLUSION Postoperative residual glandular tissue is an important tool for quantifying the risk of developing breast cancer after risk-reducing mastectomy. Different effects on residual glandular tissue were shown for the independent variables breast density, skin flap, surgical method, preoperative breast volume, and surgeon experience, so these should be considered in future surgical procedures preoperatively as well as postoperatively. Breast MRI has proven to be a suitable method to analyze the skin flap as well as the RGT.
Collapse
Affiliation(s)
- Frederic Dietzel
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Leoni Kolberg
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
- Department of Obstetrics and Gynecology, Agaplesion Bethesda Krankenhaus Wuppertal, 42109 Wuppertal, Germany
| | - Anne Sophie Vesper
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Jürgen Hoffmann
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | | | - Karin Zwiefel
- Breast Center, Kliniken der Stadt Köln, 51067 Köln, Germany
| | - Verena Friebe
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Lino M. Sawicki
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Nils Martin Bruckmann
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Kai Jannusch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Janna Morawitz
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Tanja Natascha Fehm
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Julian Kirchner
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| | - Svjetlana Mohrmann
- Department of Obstetrics and Gynecology, Medical Faculty, University Dusseldorf, 40225 Dusseldorf, Germany
| |
Collapse
|
12
|
Qualitative analysis of shared decision-making for chemoprevention in the primary care setting: provider-related barriers. BMC Med Inform Decis Mak 2022; 22:208. [PMID: 35927732 PMCID: PMC9354269 DOI: 10.1186/s12911-022-01954-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chemoprevention with anti-estrogens, such as tamoxifen, raloxifene or aromatase inhibitors, have been shown to reduce breast cancer risk in randomized controlled trials; however, uptake among women at high-risk for developing breast cancer remains low. The aim of this study is to identify provider-related barriers to shared decision-making (SDM) for chemoprevention in the primary care setting. Methods Primary care providers (PCPs) and high-risk women eligible for chemoprevention were enrolled in a pilot study and a randomized clinical trial of web-based decision support tools to increase chemoprevention uptake. PCPs included internists, family practitioners, and gynecologists, whereas patients were high-risk women, age 35–75 years, who had a 5-year invasive breast cancer risk ≥ 1.67%, according to the Gail model. Seven clinical encounters of high-risk women and their PCPs who were given access to these decision support tools were included in this study. Audio-recordings of the clinical encounters were transcribed verbatim and analyzed using grounded theory methodology. Results Six primary care providers, of which four were males (mean age 36 [SD 6.5]) and two were females (mean age 39, [SD 11.5]) and seven racially/ethnically diverse high-risk female patients participated in this study. Qualitative analysis revealed three themes: (1) Competing demands during clinical encounters; (2) lack of knowledge among providers about chemoprevention; and (3) limited risk communication during clinical encounters. Conclusions Critical barriers to SDM about chemoprevention were identified among PCPs. Providers need education and resources through decision support tools to engage in risk communication and SDM with their high-risk patients, and to gain confidence in prescribing chemoprevention in the primary care setting.
Supplementary Information The online version contains supplementary material available at 10.1186/s12911-022-01954-y.
Collapse
|
13
|
Study of Stability, Cytotoxic and Antimicrobial Activity of Chios Mastic Gum Fractions (Neutral, Acidic) after Encapsulation in Liposomes. Foods 2022; 11:foods11030271. [PMID: 35159423 PMCID: PMC8834444 DOI: 10.3390/foods11030271] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Mastic gum is a resinous sap produced by Pistacia lentiscus growing in the island of Chios (Greece) and has been recognized since Antiquity for its distinctive aroma as well as medical properties (antimicrobial, antioxidant, anti-inflammatory ones). The oral absorption of Chios Mastic gum (an insoluble polymer of poly-β-myrcene is among the most abundant contents) is poor due to its low water-solubility. We report in this study, two different Chios mastic gum extracts, the acidic mastic gum extract—AMGE—and the neutral one—NMGE, both prepared after removal of the contained polymer in order to ameliorate solubility and enhance in vivo activity. Liposomes are presented as a promising delivery system due to their physicochemical and biophysical properties to increase stability and absorption efficiency of the mastic gum extracts within the gastrointestinal (GI) tract. The aim of this study was to evaluate the stability in GI simulated conditions together with cytotoxic and antimicrobial activity of the two extracts (AMGE and NMGE) after encapsulation in a well characterized liposome formulation. Liposomes-AMGE complex showed an improved stability behavior in GI simulated conditions. Both assayed extracts showed significant dose dependent inhibition against the growth of liver cancer HepG2 cells and an interesting antimicrobial activity against several microorganisms. Conclusively, encapsulation could be evaluated as a beneficial procedure for further applications of mastic resin.
Collapse
|
14
|
Jahan N, Jones C, Rahman RL. Endocrine prevention of breast cancer. Mol Cell Endocrinol 2021; 530:111284. [PMID: 33882282 DOI: 10.1016/j.mce.2021.111284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/04/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023]
Abstract
Breast cancer (BC) is the most common non-cutaneous malignancy among women worldwide and is a significant cause of morbidity, mortality, and national health care expenditure. Unfortunately, with few exceptions like alcohol consumption, obesity, and physical activity, most BC risk factors are unmodifiable. Antiestrogen endocrine therapy, commonly known as BC chemoprevention, is an effective method of BC prevention. In multiple randomized trials, two selective estrogen receptor modulators - tamoxifen and raloxifene, and two aromatase inhibitors - exemestane and anastrozole have reduced BC incidence by 50%-65% in high-risk women. An estimated 15% of the US women between 35 and 79 years of age may qualify as high risk for BC, yet a small percentage of these women will ever have a formal BC risk assessment or a discussion of endocrine prevention options. The etiology of underutilization of endocrine prevention of BC is multifactorial - infrequent use of BC risk assessment tools in the primary care settings, insufficient knowledge of BC risk assessment tools and antiestrogen agents among primary care providers, concerns of side effects, inadequate time for counseling during primary care visit, and lack of predictive biomarkers may play significant roles. Many small studies incorporating risk assessment tools and decision-making aids showed minimal success in enhancing endocrine prevention. One critical factor for underutilization of endocrine prevention is low uptake of endocrine prevention by high-risk women even when appropriately recommended. Furthermore, adherence to BC endocrine prevention is unsatisfactorily low. Despite the current infrequent usage, endocrine prevention has the potential to reduce the public health burden of BC significantly. Innovative approaches like finding new agents, alternative dosing and schedule of currently available agents, transdermal medication delivery, increased public and professional awareness, and policymakers' commitments may bring the desired changes.
Collapse
Affiliation(s)
- Nusrat Jahan
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, Tx, 79430, USA.
| | - Catherine Jones
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, Tx, 79430, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, Tx, 79430, USA
| |
Collapse
|
15
|
Wernli KJ, Knerr S, Li T, Leppig K, Ehrlich K, Farrell D, Gao H, Bowles EJA, Graham AL, Luta G, Jayasekera J, Mandelblatt JS, Schwartz MD, O’Neill SC. Effect of Personalized Breast Cancer Risk Tool on Chemoprevention and Breast Imaging: ENGAGED-2 Trial. JNCI Cancer Spectr 2021; 5:pkaa114. [PMID: 33554037 PMCID: PMC7853161 DOI: 10.1093/jncics/pkaa114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/14/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Background Limited evidence exists about how to communicate breast density-informed breast cancer risk to women at elevated risk to motivate cancer prevention. Methods We conducted a randomized controlled trial evaluating a web-based intervention incorporating personalized breast cancer risk, information on chemoprevention, and values clarification on chemoprevention uptake vs active control. Eligible women aged 40-69 years with normal mammograms and elevated 5-year breast cancer risk were recruited from Kaiser Permanente Washington from February 2017 to May 2018. Chemoprevention uptake was measured as any prescription for raloxifene or tamoxifen within 12 months from baseline in electronic health record pharmacy data. Secondary outcomes included breast magnetic resonance imaging (MRI), mammography use, self-reported distress, and communication with providers. We calculated unadjusted odds ratios (ORs) using logistic regression models and mean differences using analysis of covariance models with 95% confidence intervals (CIs) with generalized estimating equations. Results We randomly assigned 995 women to the intervention arm (n = 492) or control arm (n = 503). The intervention (vs control) had no effect on chemoprevention uptake (OR = 1.04, 95% CI = 0.07 to 16.62). The intervention increased breast MRI use (OR = 5.65, 95% CI = 1.61 to 19.74) while maintaining annual mammography (OR = 0.98, 95% CI = 0.75 to 1.28). Women in the intervention (vs control) arm had 5.67-times higher odds of having discussed chemoprevention or breast MRI with provider by 6 weeks (OR = 5.67, 95% CI = 2.47 to 13.03) and 2.36-times higher odds by 12 months (OR = 2.36, 95% CI = 1.65 to 3.37). No measurable differences in distress were detected. Conclusions A web-based, patient-level intervention activated women at elevated 5-year breast cancer risk to engage in clinical discussions about chemoprevention, but uptake remained low.
Collapse
Affiliation(s)
- Karen J Wernli
- Correspondence to: Karen J. Wernli, PhD, Kaiser Permanente Washington Health Research Institute, 1730 Minor Ave, Suite 1600, Seattle, WA 98101, USA (e-mail: )
| | - Sarah Knerr
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Tengfei Li
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington, DC, USA
| | | | - Kelly Ehrlich
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | - Hongyuan Gao
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Erin J A Bowles
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Amanda L Graham
- Truth Initiative, Washington, DC, USA,Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - George Luta
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington, DC, USA
| | - Jinani Jayasekera
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Jeanne S Mandelblatt
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Marc D Schwartz
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Suzanne C O’Neill
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
16
|
Di Sotto A, Mancinelli R, Gullì M, Eufemi M, Mammola CL, Mazzanti G, Di Giacomo S. Chemopreventive Potential of Caryophyllane Sesquiterpenes: An Overview of Preliminary Evidence. Cancers (Basel) 2020; 12:E3034. [PMID: 33081075 PMCID: PMC7603190 DOI: 10.3390/cancers12103034] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Chemoprevention is referred to as a strategy to inhibit, suppress, or reverse tumor development and progression in healthy people along with high-risk subjects and oncologic patients through using pharmacological or natural substances. Numerous phytochemicals have been widely described in the literature to possess chemopreventive properties, although their clinical usefulness remains to be defined. Among them, caryophyllane sesquiterpenes are natural compounds widely occurring in nature kingdoms, especially in plants, fungi, and marine environments. Several structures, characterized by a common caryophyllane skeleton with further rearrangements, have been identified, but those isolated from plant essential oils, including β-caryophyllene, β-caryophyllene oxide, α-humulene, and isocaryophyllene, have attracted the greatest pharmacological attention. Emerging evidence has outlined a complex polypharmacological profile of caryophyllane sesquiterpenes characterized by blocking, suppressing, chemosensitizing, and cytoprotective properties, which suggests a possible usefulness of these natural substances in cancer chemoprevention for both preventive and adjuvant purposes. In the present review, the scientific knowledge about the chemopreventive properties of caryophyllane sesquiterpenes and the mechanisms involved have been collected and discussed; moreover, possible structure-activity relationships have been highlighted. Although further high-quality studies are required, the promising preclinical findings and the safe pharmacological profile encourage further studies to define a clinical usefulness of caryophyllane sesquiterpenes in primary, secondary, or tertiary chemoprevention.
Collapse
Affiliation(s)
- Antonella Di Sotto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Romina Mancinelli
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (R.M.); (C.L.M.)
| | - Marco Gullì
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Margherita Eufemi
- Department of Biochemical Science “A. Rossi Fanelli”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| | - Caterina Loredana Mammola
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (R.M.); (C.L.M.)
| | - Gabriela Mazzanti
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Silvia Di Giacomo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| |
Collapse
|
17
|
Fu B, Dang M, Tao J, Li Y, Tang Y. Mesoporous platinum nanoparticle-based nanoplatforms for combined chemo-photothermal breast cancer therapy. J Colloid Interface Sci 2020; 570:197-204. [DOI: 10.1016/j.jcis.2020.02.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022]
|
18
|
Pharmacological Modulation of Steroid Activity in Hormone-Dependent Breast and Prostate Cancers: Effect of Some Plant Extract Derivatives. Int J Mol Sci 2020; 21:ijms21103690. [PMID: 32456259 PMCID: PMC7279356 DOI: 10.3390/ijms21103690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/24/2022] Open
Abstract
The great majority of breast and prostate tumors are hormone-dependent cancers; hence, estrogens and androgens can, respectively, drive their developments, making it possible to use pharmacological therapies in their hormone-dependent phases by targeting the levels of steroid or modulating their physiological activity through their respective nuclear receptors when the tumors relapse. Unfortunately, at some stage, both breast and prostate cancers become resistant to pharmacological treatments that aim to block their receptors, estrogen (ER) or androgen (AR) receptors, respectively. So far, antiestrogens and antiandrogens used in clinics have been designed based on their structural analogies with natural hormones, 17-β estradiol and dihydrotestosterone. Plants are a potential source of drug discovery and the development of new pharmacological compounds. The aim of this review article is to highlight the recent advances in the pharmacological modulation of androgen or estrogen levels, and their activity through their cognate nuclear receptors in prostate or breast cancer and the effects of some plants extracts.
Collapse
|
19
|
Role of Phytochemicals in Cancer Prevention. Int J Mol Sci 2019; 20:ijms20204981. [PMID: 31600949 PMCID: PMC6834187 DOI: 10.3390/ijms20204981] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
The use of synthetic, natural, or biological agents to minimize the occurrence of cancer in healthy individuals is defined as cancer chemoprevention. Chemopreventive agents inhibit the development of cancer either by impeding DNA damage, which leads to malignancy or by reversing or blocking the division of premalignant cells with DNA damage. The benefit of this approach has been demonstrated in clinical trials of breast, prostate, and colon cancer. The continuous increase in cancer cases, failure of conventional chemotherapies to control cancer, and excessive toxicity of chemotherapies clearly demand an alternative approach. The first trial to show benefit of chemoprevention was undertaken in breast cancer patients with the use of tamoxifen, which demonstrated a significant decrease in invasive breast cancer. The success of using chemopreventive agents for protecting the high risk populations from cancer indicates that the strategy is rational and promising. Dietary components such as capsaicin, cucurbitacin B, isoflavones, catechins, lycopenes, benzyl isothiocyanate, phenethyl isothiocyanate, and piperlongumine have demonstrated inhibitory effects on cancer cells indicating that they may serve as chemopreventive agents. In this review, we have addressed the mechanism of chemopreventive and anticancer effects of several natural agents.
Collapse
|